Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 45
Filter
Add more filters










Publication year range
1.
Am J Physiol Endocrinol Metab ; 319(3): E494-E508, 2020 09 01.
Article in English | MEDLINE | ID: mdl-32691632

ABSTRACT

Hydroxysteroid 17ß dehydrogenase 12 (HSD17B12) is suggested to be involved in the elongation of very long chain fatty acids. Previously, we have shown a pivotal role for the enzyme during mouse development. In the present study we generated a conditional Hsd17b12 knockout (HSD17B12cKO) mouse model by breeding mice homozygous for a floxed Hsd17b12 allele with mice expressing the tamoxifen-inducible Cre recombinase at the ROSA26 locus. Gene inactivation was induced by administering tamoxifen to adult mice. The gene inactivation led to a 20% loss of body weight within 6 days, associated with drastic reduction in both white (83% males, 75% females) and brown (65% males, 60% females) fat, likely due to markedly reduced food and water intake. Furthermore, the knockout mice showed sickness behavior and signs of liver toxicity, specifically microvesicular hepatic steatosis and increased serum alanine aminotransferase (4.6-fold in males, 7.7-fold in females). The hepatic changes were more pronounced in females than males. Proinflammatory cytokines, such as interleukin-6 (IL-6), IL-17, and granulocyte colony-stimulating factor, were increased in the HSD17B12cKO mice indicating an inflammatory response. Serum lipidomics study showed an increase in the amount of dihydroceramides, despite the dramatic overall loss of lipids. In line with the proposed role for HSD17B12 in fatty acid elongation, we observed accumulation of ceramides, dihydroceramides, hexosylceramides, and lactosylceramides with shorter than 18-carbon fatty acid side chains in the serum. The results indicate that HSD17B12 is essential for proper lipid homeostasis and HSD17B12 deficiency rapidly results in fatal systemic inflammation and lipolysis in adult mice.


Subject(s)
17-Hydroxysteroid Dehydrogenases/physiology , Homeostasis/physiology , 17-Hydroxysteroid Dehydrogenases/genetics , Adipose Tissue, Brown/drug effects , Adipose Tissue, Brown/metabolism , Adipose Tissue, White/drug effects , Adipose Tissue, White/metabolism , Animals , Behavior, Animal , Body Weight/genetics , Cytokines/metabolism , Fatty Acids/metabolism , Feeding Behavior , Female , Homeostasis/genetics , Lipid Metabolism/genetics , Lipid Metabolism/physiology , Lipidomics , Liver Diseases/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Sex Characteristics , Tamoxifen/pharmacology
2.
FASEB J ; 34(5): 6111-6128, 2020 05.
Article in English | MEDLINE | ID: mdl-32190925

ABSTRACT

Hydroxysteroid (17ß) dehydrogenase type 3 (HSD17B3) deficiency causes a disorder of sex development in humans, where affected males are born with female-appearing external genitalia, but are virilized during puberty. The hormonal disturbances observed in the Hsd17b3 knockout mice (HSD17B3KO), generated in the present study, mimic those found in patients with HSD17B3 mutations. Identical to affected humans, serum T in the adult HSD17B3KO mice was within the normal range, while a striking increase was detected in serum A-dione concentration. This resulted in a marked reduction of the serum T/A-dione ratio, a diagnostic hallmark for the patients with HSD17B3 deficiency. However, unlike humans, male HSD17B3KO mice were born with normally virilized phenotype, but presenting with delayed puberty. In contrast to the current belief, data from HSD17B3KO mice show that the circulating T largely originates from the testes, indicating a strong compensatory mechanism in the absence of HSD17B3. The lack of testicular malignancies in HSD17B3KO mice supports the view that testis tumors in human patients are due to associated cryptorchidism. The HSD17B3KO mice presented also with impaired Leydig cell maturation and signs of undermasculinization in adulthood. The identical hormonal disturbances between HSD17B3 deficient knockout mice and human patients make the current mouse model valuable for understanding the mechanism of the patient phenotypes, as well as endocrinopathies and compensatory steroidogenic mechanisms in HSD17B3 deficiency.


Subject(s)
17-Hydroxysteroid Dehydrogenases/physiology , Gonadal Steroid Hormones/blood , Infertility, Male/pathology , Leydig Cells/pathology , Mutation , 17-Hydroxysteroid Dehydrogenases/deficiency , 17-Hydroxysteroid Dehydrogenases/genetics , Animals , Female , Infertility, Male/etiology , Leydig Cells/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout
3.
Anim Sci J ; 89(1): 158-166, 2018 Jan.
Article in English | MEDLINE | ID: mdl-28877400

ABSTRACT

The enzyme 3ß-hydroxysteroid dehydrogenase (3ß-HSD) plays an important role in androstenone metabolism in pig liver, and its defective expression is related to the development of boar taint. Early age castration is a common practice in many countries to avoid boar taint, yet whether and how castration affects porcine hepatic 3ß-HSD expression are still poorly understood. In this study, we aimed to compare the expression of 3ß-HSD between intact (boars) and castrated (barrows) male pigs, and to explore the potential factors regulating 3ß-HSD transcription. Compared to barrows, boars showed worse carcass quality. Boars had significantly higher levels of serum androstenone (P < 0.01), testosterone (P < 0.01) and hepatic cortisol (P < 0.05), which were contrary to significantly lower expression of 3ß-HSD messenger RNA (P < 0.01) and protein (P < 0.01) in the liver. Significant differences were detected for the hepatic expression of androgen receptor (AR) and CCAAT/enhancer binding protein ß (C/EBPß). Chromatin immunoprecipitation (ChIP) assay demonstrated reduced histone H3 acetylation (P < 0.05) but increased glucocorticoid receptor (GR) binding to 3ß-HSD gene promoter in boars (P < 0.05). These results indicate that GR binding to 3ß-HSD promoter is involved in the differential hepatic 3ß-HSD expression between boars and barrows.


Subject(s)
17-Hydroxysteroid Dehydrogenases/genetics , 17-Hydroxysteroid Dehydrogenases/metabolism , Castration , Gene Expression , Receptors, Glucocorticoid/metabolism , Swine/genetics , Swine/metabolism , 17-Hydroxysteroid Dehydrogenases/physiology , Acetylation , Androsterone/metabolism , Animals , CCAAT-Enhancer-Binding Protein-beta/metabolism , Histones/metabolism , Hydrocortisone/metabolism , Liver/metabolism , Male , Promoter Regions, Genetic , Protein Binding , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Androgen/metabolism , Testosterone/metabolism
5.
Horm Metab Res ; 44(13): 949-56, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22864907

ABSTRACT

17ßHSD enzymes catalyze the stereospecific oxidation/reduction at carbon 17ß of androgens and estrogens, and are important players in intracrine sex hormone synthesis. The biological relevance of 17ßHSD14, first named retSDR3, is largely unknown. We generated and validated an antibody targeting the 17ßHSD14 antigen and used this for immunohistochemical evaluation of expression patterns in 33 healthy human tissues. Furthermore, sex steroid conversional activity in HSD17B14 overexpressing HEK293 and MCF10A cells was investigated by assessing interconversion products of estrone, estradiol, androstenedione, testosterone, and dehydroepiandrosterone. Immunohistochemical staining patterns of 17ßHSD14 with the enzyme being primarily expressed in glandular epithelial tissue reveal an enzyme with possible implications in the secretion or conversion of externally derived compounds. A role for 17ßHSD14 in sex steroid metabolism is supported by the finding that 17HSD14 oxidizes both estradiol and testosterone into less bioactive steroid metabolites estrone and androstenedione, respectively.


Subject(s)
17-Hydroxysteroid Dehydrogenases/analysis , 17-Hydroxysteroid Dehydrogenases/physiology , Antibody Specificity , HEK293 Cells , Humans , Immunohistochemistry
6.
Endocrinology ; 151(8): 3514-20, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20534728

ABSTRACT

Prostate cancer usually responds to androgen deprivation therapy, although the response in metastatic disease is almost always transient and tumors eventually progress as castration-resistant prostate cancer (CRPC). CRPC continues to be driven by testosterone or dihydrotestosterone from intratumoral metabolism of 19-carbon adrenal steroids from circulation, and/or de novo intratumoral steroidogenesis. Both mechanisms require 3beta-hydroxysteroid dehydrogenase (3betaHSD) metabolism of Delta(5)-steroids, including dehydroepiandrosterone (DHEA) and Delta(5)-androstenediol (A5diol), to testosterone. In contrast, reports that DHEA and A5diol directly activate the androgen receptor (AR) suggest that 3betaHSD metabolism is not required and that 3betaHSD inhibitors would be ineffective in the treatment of CRPC. We hypothesized that activation of AR in prostate cancer by DHEA and A5diol requires their conversion via 3betaHSD to androstenedione and testosterone, respectively. Here, we show that DHEA and A5diol induce AR chromatin occupancy and AR-regulated genes. Furthermore, we show that Delta(5)-androgens undergo 3beta-dehydrogenation in prostate cancer and that induction of AR nuclear translocation, AR chromatin occupancy, transcription of PSA, TMPRSS2, and FKBP5, as well as cell proliferation by DHEA and A5diol, are all blocked by inhibitors of 3betaHSD. These findings demonstrate that DHEA and A5diol must be metabolized by 3betaHSD to activate AR in these models of CRPC. Furthermore, this work suggests that 3betaHSD may be exploited as a pharmacologic target in the treatment of CRPC.


Subject(s)
17-Hydroxysteroid Dehydrogenases/antagonists & inhibitors , Antineoplastic Agents/administration & dosage , Carcinoma/drug therapy , Prostatic Neoplasms/drug therapy , 17-Hydroxysteroid Dehydrogenases/metabolism , 17-Hydroxysteroid Dehydrogenases/physiology , Antineoplastic Agents/pharmacology , Carcinoma/genetics , Carcinoma/pathology , Carcinoma/surgery , Cell Line, Tumor , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Dehydroepiandrosterone/metabolism , Dihydrotestosterone/administration & dosage , Dihydrotestosterone/analogs & derivatives , Dihydrotestosterone/pharmacology , Dose-Response Relationship, Drug , Drug Delivery Systems , Drug Evaluation, Preclinical , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/pharmacology , Humans , Male , Models, Biological , Orchiectomy , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Prostatic Neoplasms/surgery , Receptors, Androgen/metabolism , Treatment Failure
7.
Biochim Biophys Acta ; 1781(11-12): 694-702, 2008.
Article in English | MEDLINE | ID: mdl-18773970

ABSTRACT

Mice with inactivation of the D-specific multifunctional protein 2 (MFP2), a crucial enzyme of peroxisomal beta-oxidation, develop multiple pathologies in diverse tissues already starting in the postnatal period. Gene expression profiling performed on liver of 2-day-old pups revealed up-regulation of PPAR alpha responsive genes in knockout mice. Surprisingly, also genes involved in cholesterol biosynthesis were markedly induced. Real-time PCR confirmed the induction of PPAR alpha target genes and of HMGCR and SREBP2, both involved in cholesterol synthesis, in lactating and in adult MFP2 knockout mice. In accordance, the rate of cholesterol biosynthesis was significantly increased in liver of knockout mice but the hepatic cholesterol concentration was unaltered. In MFP2/PPAR alpha double knockout mice, up-regulations of SREBP2 and HMGCR were markedly attenuated. These data demonstrate a tight interrelationship between induction of PPAR alpha by endogenous ligands and up-regulation of genes of cholesterol biosynthesis through increased expression of SREBP2.


Subject(s)
17-Hydroxysteroid Dehydrogenases/physiology , Disease Models, Animal , Enoyl-CoA Hydratase/physiology , Liver/metabolism , Multienzyme Complexes/physiology , PPAR alpha/biosynthesis , Sterol Regulatory Element Binding Protein 2/biosynthesis , Animals , Blotting, Western , Cells, Cultured , Cholesterol/biosynthesis , Gene Expression Profiling , Gene Expression Regulation , Hepatocytes/cytology , Hepatocytes/metabolism , Lactation , Mice , Mice, Knockout , Oligonucleotide Array Sequence Analysis , PPAR alpha/genetics , Peroxisomal Multifunctional Protein-2 , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Sterol Regulatory Element Binding Protein 2/genetics , Up-Regulation
8.
J Endocrinol Invest ; 31(1): 85-91, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18296911

ABSTRACT

The 17beta-hydroxysteroid dehydrogenases (17betaHSD) gene family comprises different enzymes involved in the biosynthesis of active steroid hormones. The 17betaHSD type 3 (17betaHSD3) isoenzyme catalyzes the reductive conversion of the inactive C19-steroid, Delta4-androstenedione (Delta4- A), into the biologically active androgen, testosterone (T), in the Leydig cells of the testis. It is encoded by the 17beta-hydroxysteroid dehydrogenase type 3 (HSD17B3) gene, which maps to chromosome 9q22. Mutations in the HSD17B3 gene are associated with a rare form of 46,XY disorder of sex development referred to as 17betaHSD3 deficiency (or as 17-ketosteroid reductase deficiency), due to impaired testicular conversion of Delta4-A into T. 46,XY patients with 17betaHSD3 deficiency are usually classified as female at birth, raised as such, but develop secondary male features at puberty. Diagnosis, and consequently early treatment, is difficult because clinical signs from birth until puberty may be mild or absent. Biochemical diagnosis of 17betaHSD3 deficiency requires measurement of serum T/Delta4-A ratio after hCG stimulation test in pre-pubertal subjects, while baseline values seem to be informative in early infancy and adolescence. However, low basal T/Delta4-A ratio is not specific for 17betaHSD3 deficiency, being sometimes also found in patients with other defects in T synthesis or with Leydig cells hypoplasia. Mutational analysis of the 17HSDB3 gene is useful in confirming the clinical diagnosis of 17betaHSD3 deficiency. This review describes clinical findings, diagnosis, and molecular basis of this rare disease.


Subject(s)
17-Hydroxysteroid Dehydrogenases/genetics , Disorders of Sex Development/diagnosis , Disorders of Sex Development/genetics , Endocrine System/physiology , 17-Hydroxysteroid Dehydrogenases/deficiency , 17-Hydroxysteroid Dehydrogenases/physiology , Amino Acid Sequence , Disorders of Sex Development/physiopathology , Disorders of Sex Development/therapy , Female , Gender Identity , Humans , Male , Molecular Diagnostic Techniques , Mutation , Testis/enzymology
9.
Breast Cancer Res Treat ; 111(2): 191-202, 2008 Sep.
Article in English | MEDLINE | ID: mdl-17934808

ABSTRACT

There is a large and compelling body of epidemiological and experimental evidence that oestrogens are instrumental in the aetiology of breast cancer. Their mechanisms of action are varied, including stimulation of cellular proliferation through receptor-mediated hormonal activity, increasing genetic mutation rates through cytochrome P450-mediated metabolic activation, and induction of aneuploidy. The local biosynthesis of oestrogens especially in postmenopausal women is believed to play a very important role in the pathogenesis and development of hormone dependent breast carcinoma and the over-expression of regulatory enzymes seems to be associated with the development of a more aggressive disease and associated with poor outcome and increased local and distant recurrences. In this article we highlight the role of CYP19 gene expression and aromatase activity in mammary carcinogenesis. Other oestrogen producing (17-beta-hydroxysteroid dehydrogenase and steroid sulphatase) and catalyzing enzymes (3-beta-hydroxysteroid dehydrogenase, Oestrogen sulfotransferase, CYP1A1, CYP1B1, and CYP3A4) are also discussed in some detail. Understanding the mechanisms that regulate these enzymes is crucial to the development of new endocrine therapies in post-menopausal females with hormone dependant breast cancer. Currently, third generation aromatase inhibitors has revolutionized the treatment of oestrogen dependant breast cancer. However, the important role of both STS and 17-beta-HSD type 1 in local oestrogen production provides novel potential targets for endocrine therapy. Such endocrine therapy is currently being explored and the development of STS inhibitors, combined aromatase/steroid sulfatase inhibitors and 17-beta-HSD type 1 inhibitors is underway with promising initial results.


Subject(s)
17-Hydroxysteroid Dehydrogenases/physiology , Aromatase/physiology , Breast Neoplasms/etiology , Estrogens/biosynthesis , Steryl-Sulfatase/physiology , 17-Hydroxysteroid Dehydrogenases/antagonists & inhibitors , 17-Hydroxysteroid Dehydrogenases/genetics , Animals , Aromatase/genetics , Aromatase Inhibitors/therapeutic use , Breast Neoplasms/drug therapy , Breast Neoplasms/enzymology , Enzyme Inhibitors/therapeutic use , Humans , Steryl-Sulfatase/antagonists & inhibitors , Steryl-Sulfatase/genetics
10.
Reprod Biol Endocrinol ; 5: 30, 2007 Jul 10.
Article in English | MEDLINE | ID: mdl-17623101

ABSTRACT

BACKGROUND: During human pregnancy, the placental villi produces high amounts of estradiol. This steroid is secreted by the syncytium, which is directly in contact with maternal blood. Estradiol has to cross placental foetal vessels to reach foetal circulation. The enzyme 17beta-hydroxysteroid dehydrogenase type 2 (17beta-HSD2) was detected in placental endothelial cells of foetal vessels inside the villi. This enzyme catalyzes the conversion of estradiol to estrone, and of testosterone to androstenedione. It was proposed that estradiol level into foetal circulation could be regulated by 17beta-HSD2. METHODS: We obtained placentas from 10 to 26 6/7 weeks of pregnancy from women undergoing voluntary termination of pregnancy, term placentas were collected after normal spontaneous vaginal deliveries. We quantified 17beta-HSD2 mRNA levels in mid-gestation and term human placenta by RT-QPCR. We produced a new anti-17beta-HSD2 antibody to study its spatio-temporal expression by immunohistochemistry. We also compared steroid levels (testosterone, estrone and estradiol) and 17beta-HSD2 mRNA and protein levels between term placenta and endometrium. RESULTS: High 17beta-HSD2 mRNA and protein levels were found in both mid-gestation and term placentas. However, we showed that 17beta-HSD2 mRNA levels increase by 2.27 fold between mid-gestation and term. This period coincides with a transitional phase in the development of the villous vasculature. In mid-gestation placenta, high levels of 17beta-HSD2 were found in mesenchymal villi and immature intermediate villi, more precisely in endothelial cells of the stromal channel. At term, high levels of 17beta-HSD2 were found in the numerous sinusoidal capillaries of terminal villi. 17beta-HSD2 mRNA and protein levels in term placentas were respectively 25.4 fold and 30 to 60 fold higher than in the endometrium. Steroid levels were also significantly higher in term placenta than in the endometrium. CONCLUSION: The spatial and temporal expression of 17beta-HSD2 in the placenta during pregnancy and the comparison of 17beta-HSD2 expression and steroid levels between placental villi and endometrium are compatible with a role in the modulation of active and inactive forms of estrogens. Our observations strongly support the hypothesis that 17beta-HSD2 acts as a barrier decreasing estradiol secretion rates in the foetal circulation.


Subject(s)
17-Hydroxysteroid Dehydrogenases/metabolism , Chorionic Villi/enzymology , Pregnancy Trimester, Second/genetics , Pregnancy Trimester, Third/genetics , RNA, Messenger/biosynthesis , 17-Hydroxysteroid Dehydrogenases/biosynthesis , 17-Hydroxysteroid Dehydrogenases/genetics , 17-Hydroxysteroid Dehydrogenases/physiology , Chorionic Villi/physiology , Estradiol Dehydrogenases , Female , Gene Expression Regulation, Enzymologic , Humans , Pregnancy , RNA, Messenger/analysis , RNA, Messenger/genetics
11.
J Steroid Biochem Mol Biol ; 104(3-5): 334-9, 2007 May.
Article in English | MEDLINE | ID: mdl-17467981

ABSTRACT

Determining the functional aspects of a gene or protein is a difficult and time-consuming process. De novo analysis is surely the hardest and so it is often quite useful to start with a comparison to functionally or structurally related proteins. Although 17beta-hydroxysteroid dehydrogenase type 1 (17beta-HSD 1) can hardly be called a new protein but rather the best characterized among the family of 17beta-HSDs some aspects of structure-function relationships remain unclear. We have sought new aspects of 17beta-HSD 1 function through a comparison with its closest homolog, a photoreceptor-associated retinol dehydrogenase (prRDH). Overall amino acid identity and size of the proteins are highly conserved, but major differences occur in the C-termini, where prRDH, but not 17beta-HSD 1, harbors motifs indicative of membrane localization. To gain insight into substrate discrimination by prRDH and 17beta-HSD 1, we constructed 3D-structure models of the corresponding zebrafish enzymes. Investigation of the substrate binding site revealed a few identical amino acids, and suggested a role for G143 in zebrafish 17beta-HSD 1 and M146 and M147 in the two zebrafish paralogs prRDH 1 and prRDH 2, respectively, in substrate specificity. Activity measurements of modified proteins in transiently transfected intact HEK 293 cells hint at a putative role of these amino acids in discrimination between steroid and retinoid substrates.


Subject(s)
17-Hydroxysteroid Dehydrogenases/physiology , Alcohol Oxidoreductases/physiology , 17-Hydroxysteroid Dehydrogenases/genetics , Alcohol Oxidoreductases/genetics , Amino Acid Sequence , Animals , Cells, Cultured , Humans , Models, Molecular , Molecular Sequence Data , Protein Structure, Tertiary , Retinoids/metabolism , Sequence Alignment , Sequence Homology, Amino Acid , Steroids/metabolism , Substrate Specificity , Transfection , Zebrafish/genetics
12.
Biochim Biophys Acta ; 1761(9): 973-94, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16766224

ABSTRACT

The mammalian multifunctional protein-2 (MFP-2, also called multifunctional enzyme 2, D-bifunctional enzyme or 17-beta-estradiol dehydrogenase type IV) was identified by several groups about a decade ago. It plays a central role in peroxisomal beta-oxidation as it handles most, if not all, peroxisomal beta-oxidation substrates. Deficiency of this enzyme in man causes a severe developmental syndrome with abnormalities in several organs but in particular in the brain, leading to death within the first year of life. Accumulation of branched-long-chain fatty acids and very-long-chain fatty acids and a disturbed synthesis of bile acids were documented in these patients. A mouse model with MFP-2 deficiency only partly phenocopies the human disease. Although the expected metabolic abnormalities are present, no neurodevelopmental aberrations are observed. However, the survival of these mice into adulthood allowed to document the importance of this enzyme for the normal functioning of the brain, eyes and testis. In the present review, the identification and biochemical characteristics of MFP-2, and the consequences of MFP-2 dysfunction in humans and in mice will be discussed.


Subject(s)
17-Hydroxysteroid Dehydrogenases/physiology , Enoyl-CoA Hydratase/physiology , Models, Molecular , Multienzyme Complexes/physiology , Peroxisomes/enzymology , 17-Hydroxysteroid Dehydrogenases/chemistry , 17-Hydroxysteroid Dehydrogenases/deficiency , Abnormalities, Multiple/enzymology , Abnormalities, Multiple/pathology , Amino Acid Sequence , Animals , Brain/abnormalities , Brain/growth & development , Enoyl-CoA Hydratase/chemistry , Enoyl-CoA Hydratase/deficiency , Eye Abnormalities/enzymology , Eye Abnormalities/pathology , Fatty Acids/metabolism , Humans , Lipid Metabolism , Male , Mice , Mice, Knockout , Molecular Sequence Data , Multienzyme Complexes/chemistry , Multienzyme Complexes/deficiency , Organ Specificity , Peroxisomal Multifunctional Protein-2 , Testis/abnormalities , Testis/growth & development
13.
Article in English | MEDLINE | ID: mdl-16611167

ABSTRACT

Human type 10 17beta-hydroxysteroid dehydrogenase (HSD) is a homotetrameric protein located in mitochondria. This enzyme was alternatively named short chain L-3-hydroxyacyl-CoA dehydrogenase (SCHSD). This NAD(H)-dependent dehydrogenase is essential for the metabolism of branched-chain fatty acids and isoleucine, and is expressed in a variety of tissues, e.g., prostate, brain, liver, and heart. This enzyme inactivates 17beta-estradiol and exhibits a strong oxidative 3alpha-HSD activity to convert 5alpha-androstanediol and allopregnanolone into 5alpha-dihydrotestosterone (5alpha-DHT) and 5alpha-dihydroprogesterone, respectively, in living cells. Certain malignant prostatic epithelial cells and activated astrocytes in Alzheimer's disease patient's brain contain extraordinarily high levels of this enzyme. This mitochondrial dehydrogenase enables prostate cancer cells to generate 5alpha-DHT in the absence of testosterone. Its inactivation of allopregnanolone is important to the modulation of GABA(A) receptor. Among steroidogenic enzymes 17beta-HSD10 plays a significant part in the intracrinology. Although this protein has an affinity for amyloid-beta peptide, its role in the pathogenesis of Alzheimer's disease is far from clear. Additional knowledge of this versatile enzyme would provide the foundation for designing new drugs aimed at treating some neurological diseases and certain types of cancers.


Subject(s)
Fatty Acids/metabolism , Isoleucine/metabolism , 17-Hydroxysteroid Dehydrogenases/antagonists & inhibitors , 17-Hydroxysteroid Dehydrogenases/physiology , 3-Hydroxyacyl CoA Dehydrogenases/antagonists & inhibitors , 3-Hydroxyacyl CoA Dehydrogenases/physiology , Amino Acid Metabolism, Inborn Errors/metabolism , Androgens/biosynthesis , Animals , Estrogens/pharmacology , Humans , Mitochondria/enzymology , Molecular Sequence Data , Neurotransmitter Agents/metabolism
14.
Mol Cell Endocrinol ; 248(1-2): 141-8, 2006 Mar 27.
Article in English | MEDLINE | ID: mdl-16406264

ABSTRACT

Sex steroid hormone signaling regulates the development, growth, and functioning of the breast and the prostate and plays a role in the development and progression of cancer in these organs. The intracellular concentration of active sex steroid hormones in target tissues is regulated by several enzymes, including 17beta-hydroxysteroid dehydrogenases (17HSDs). Changes in the expression patterns of these enzymes may play a pathophysiological role in malignant transformation. We recently analyzed the mRNA expressions of the 17HSD type 1, 2, and 5 enzymes in about 800 breast carcinoma specimens. Both 17HSD type 1 and 2 mRNAs were detected in normal breast tissue from premenopausal women but not in specimens from postmenopausal women. The patients with tumors expressing 17HSD type 1 mRNA or protein had significantly shorter overall and disease-free survival than the other patients. The expression of 17HSD type 5 was significantly higher in breast tumor specimens than in normal tissue. Cox multivariate analyses showed that 17HSD type 1, tumor size, and estrogen receptor alpha (ERalpha) had independent prognostic significance. We developed, using a LNCaP prostate cancer cell line, a model to study the malignant transformation of prostate cancer and showed that androgen-sensitive LNCaP cells are transformed into neuroendocrine-like cells when cultured without androgens and, eventually into highly proliferating androgen-independent cells. We conducted Northern hybridizations and microarrays to analyze the gene expression during these processes. Substantial changes in the expressions of steroid metabolizing enzymes occurred during the transformation process. The variations in steroid-metabolizing enzymes during cancer progression may be crucial in the regulation of the growth and function of organs.


Subject(s)
17-Hydroxysteroid Dehydrogenases/physiology , Breast Neoplasms/enzymology , Cell Proliferation , Gonadal Steroid Hormones/metabolism , Prostatic Neoplasms/enzymology , 17-Hydroxysteroid Dehydrogenases/analysis , 17-Hydroxysteroid Dehydrogenases/genetics , Androgens/metabolism , Breast Neoplasms/pathology , Female , Humans , Male , Prostate/enzymology , Prostate/metabolism
15.
Mol Cell Endocrinol ; 248(1-2): 61-71, 2006 Mar 27.
Article in English | MEDLINE | ID: mdl-16414178

ABSTRACT

17Beta-hydroxysteroid dehydrogenases (17beta-HSDs) catalyze the NAD(P)(H) dependent oxidoreduction at C17 oxo/beta-hydroxyl groups of androgen and estrogen hormones. This reversible reaction constitutes an important pre-receptor control mechanism for nuclear receptor ligands, since the conversion "switches" between the 17beta-OH receptor ligands and their inactive 17-oxo metabolites. At present, 14 mammalian 17beta-HSDs are described, of which at least 11 exist within the human genome, encoded by different genes. The enzymes differ in their expression pattern, nucleotide cofactor preference, steroid substrate specificity and subcellular localization, and thus constitute a complex system ensuring cell-specific adaptation and regulation of sex steroid hormone levels. Broad and overlapping substrate specificities with enzymes involved in lipid metabolism suggest interactions of several 17beta-HSDs with other metabolic pathways. Several 17beta-HSDs enzymes constitute promising drug targets, of particular importance in cancer, metabolic diseases, neurodegeneration and possibly immunity.


Subject(s)
17-Hydroxysteroid Dehydrogenases/chemistry , 17-Hydroxysteroid Dehydrogenases/physiology , 17-Hydroxysteroid Dehydrogenases/drug effects , Humans , Protein Conformation , Substrate Specificity
16.
J Pharmacol Exp Ther ; 316(3): 1300-9, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16339391

ABSTRACT

Tibolone [[7alpha,17alpha]-17-hydroxy-7-methyl-19-norpregn-5(10)-en-20-yn-3-one] is used to treat climacteric symptoms and prevent osteoporosis. It exerts tissue-selective effects via site-specific metabolism into 3alpha- and 3beta-hydroxymetabolites and a Delta4-isomer. Recombinant human cytosolic aldo-keto reductases 1C1 and 1C2 (AKR1C1 and AKR1C2) produce 3beta-hydroxytibolone, and the liver-specific AKR1C4 produces predominantly 3alpha-hydroxytibolone. These observations may account for the appearance of 3beta-hydroxytibolone in target tissues and 3alpha-hydroxytibolone in the circulation. Using liver autopsy samples (which express AKR1C1-AKR1C4), tibolone was reduced via 3alpha- and 3beta-hydroxysteroid dehydrogenase (HSD) activity. 3beta-Hydroxytibolone was exclusively formed in the cytosol and was inhibited by the AKR1C2-specific inhibitor 5beta-cholanic acid-3alpha, 7alpha-diol. The cytosolic formation of 3alpha-hydroxytibolone was inhibited by an AKR1C4-selective inhibitor, phenolphthalein. The ratio of these stereoisomers was 4:1 in favor of 3beta-hydroxytibolone. In HepG2 cell cytosol and intact cells (which do not express AKR1C4), tibolone was exclusively reduced to 3beta-hydroxytibolone and was blocked by the AKR1C1-AKR1C3 inhibitor flufenamic acid. In primary hepatocytes (which express AKR1C1-AKR1C4), time-dependent reduction of tibolone into 3beta- and 3alpha-hydroxytibolone was observed again in a 4:1 ratio. 3beta-HSD activity was inhibited by both 5beta-cholanic acid-3alpha,7alpha-diol and flufenamic acid, implicating a role for AKR1C2 and AKR1C1. By contrast, the formation of 3alpha-hydroxytibolone was exclusively inhibited by phenolphthalein implicating AKR1C4 in this reaction. 3beta- and 3alpha-Hydroxytibolone were rapidly metabolized into polar metabolites (>85%). The formation of minor amounts of tibolone was also observed followed by AKR1C-catalyzed epimerization. The low hepatic formation of 3alpha-hydroxytibolone suggests that AKR1C4 is not the primary source of this metabolite and instead it maybe formed by an intestinal or enterobacterial 3alpha-HSD.


Subject(s)
17-Hydroxysteroid Dehydrogenases/physiology , 20-Hydroxysteroid Dehydrogenases/physiology , 3-Hydroxysteroid Dehydrogenases/physiology , Hydroxyprostaglandin Dehydrogenases/physiology , Hydroxysteroid Dehydrogenases/physiology , Liver/metabolism , Norpregnenes/metabolism , Oxidoreductases/physiology , Aldo-Keto Reductase Family 1 Member C3 , Bile Acids and Salts/pharmacology , Catalysis , Cells, Cultured , Flufenamic Acid/pharmacology , Hepatocytes/enzymology , Humans , Phenolphthalein/pharmacology
17.
Prostate ; 65(2): 159-70, 2005 Oct 01.
Article in English | MEDLINE | ID: mdl-15924334

ABSTRACT

BACKGROUND: Endocrine therapy of prostate cancer (PCa) relies on agents which disrupt the biosynthesis of testosterone in the testis and/or by direct antagonism of active hormone on the androgen receptor (AR) in non-gonadal target tissues of hormone action such as the prostate. METHODS: In an effort to evaluate new therapies which could inhibit gonadal or non-gonadal testosterone biosynthesis, we developed high throughput biochemical and cellular screening assays to identify inhibitors of 17beta-hydroxysteroid dehydrogenase type III (17beta-HSD3), the enzyme catalyzing the conversion of androstenedione (AdT) to testosterone. RESULTS: Initial screening efforts identified a natural product, 18beta-glycyrrhetinic acid, and a novel derivative of AdT, 3-O-benzylandrosterone, as potent inhibitors of the enzyme. Further efforts led to the identification of several classes of non-steroidal, low molecular weight compounds that potently inhibited 17beta-HSD3 enzymatic activity. One of the most potent classes of 17beta-HSD3 inhibitors was a series of anthranilamide small molecules identified from a collection of compounds related to non-steroidal modulators of nuclear hormone receptors. The anthranilamide based 17beta-HSD3 inhibitors were exemplified by BMS-856, a compound displaying low nanomolar inhibition of 17beta-HSD3 enzymatic activity. In addition, this series of compounds displayed potent inhibition of 17beta-HSD3-mediated cellular conversion of AdT to testosterone and inhibited the 17beta-HSD3-mediated conversion of testosterone necessary to promote AR-dependent transcription. CONCLUSIONS: The identification of non-steroidal functional inhibitors of 17beta-HSD3 may be a useful complementary approach for the disruption of testosterone biosynthesis in the treatment of PCa.


Subject(s)
17-Hydroxysteroid Dehydrogenases/antagonists & inhibitors , 17-Hydroxysteroid Dehydrogenases/physiology , Anti-Inflammatory Agents/pharmacology , Glycyrrhetinic Acid/pharmacology , Testosterone/biosynthesis , ortho-Aminobenzoates/pharmacology , Androstenedione/metabolism , Drug Evaluation, Preclinical , Humans , Male , Prostatic Neoplasms/drug therapy
18.
Trends Endocrinol Metab ; 16(4): 167-75, 2005.
Article in English | MEDLINE | ID: mdl-15860413

ABSTRACT

Human 17beta-hydroxysteroid dehydrogenase type 10 (17beta-HSD10) is a mitochondrial enzyme encoded by the SCHAD gene, which escapes chromosome X inactivation. 17Beta-HSD10/SCHAD mutations cause a spectrum of clinical conditions, from mild mental retardation to progressive infantile neurodegeneration. 17Beta-HSD10/SCHAD is essential for the metabolism of isoleucine and branched-chain fatty acids. It can inactivate 17beta-estradiol and steroid modulators of GABA(A) receptors, and convert 5alpha-androstanediol into 5alpha-dihydrotestosterone (DHT). Certain malignant prostatic epithelial cells contain high levels of 17beta-HSD10, generating 5alpha-DHT in the absence of testosterone. 17Beta-HSD10 has an affinity for amyloid-beta peptide, and might be linked to the mitochondrial dysfunction seen in Alzheimer's disease. This versatile enzyme might provide a new drug target for neuronal excitability control and for intervention in Alzheimer's disease and certain cancers.


Subject(s)
17-Hydroxysteroid Dehydrogenases/physiology , 17-Hydroxysteroid Dehydrogenases/genetics , 17-Hydroxysteroid Dehydrogenases/metabolism , 3-Hydroxyacyl CoA Dehydrogenases/genetics , 3-Hydroxyacyl CoA Dehydrogenases/metabolism , Alzheimer Disease/enzymology , Alzheimer Disease/etiology , Animals , Gonadal Steroid Hormones/metabolism , Humans , Isoleucine/metabolism , Neurotransmitter Agents/metabolism , Steroids/metabolism
19.
J Biol Chem ; 280(19): 18658-66, 2005 May 13.
Article in English | MEDLINE | ID: mdl-15769750

ABSTRACT

Peroxisomal beta-oxidation is an essential step in bile acid synthesis, since it is required for shortening of C27-bile acid intermediates to produce mature C24-bile acids. D-Bifunctional protein (DBP) is responsible for the second and third step of this beta-oxidation process. However, both patients and mice with a DBP deficiency still produce C24-bile acids, although C27-intermediates accumulate. An alternative pathway for bile acid biosynthesis involving the peroxisomal L-bifunctional protein (LBP) has been proposed. We investigated the role of LBP and DBP in bile acid synthesis by analyzing bile acids in bile, liver, and plasma from LBP, DBP, and LBP:DBP double knock-out mice. Bile acid biosynthesis, estimated by the ratio of C27/C24-bile acids, was more severely affected in double knock-out mice as compared with DBP-/- mice but was normal in LBP-/- mice. Unexpectedly, trihydroxycholestanoyl-CoA oxidase was inactive in double knock-out mice due to a peroxisomal import defect, preventing us from drawing any firm conclusion about the potential role of LBP in an alternative bile acid biosynthesis pathway. Interestingly, the immature C27-bile acids in DBP and double knock-out mice remained unconjugated in juvenile mice, whereas they occurred as taurine conjugates after weaning, probably contributing to the minimal weight gain of the mice during the lactation period. This correlated with a marked induction of bile acyl-CoA:amino acid N-acyltransferase expression and enzyme activity between postnatal days 10 and 21, whereas the bile acyl-CoA synthetases increased gradually with age. The nuclear receptors hepatocyte nuclear factor-4alpha, farnesoid X receptor, and peroxisome proliferator receptor alpha did not appear to be involved in the up-regulation of the transferase.


Subject(s)
17-Hydroxysteroid Dehydrogenases/physiology , 3-Hydroxyacyl CoA Dehydrogenases/physiology , Bile Acids and Salts/chemistry , Enoyl-CoA Hydratase/physiology , Gene Expression Regulation, Developmental , Isomerases/physiology , Multienzyme Complexes/physiology , 17-Hydroxysteroid Dehydrogenases/chemistry , 3-Hydroxyacyl CoA Dehydrogenases/chemistry , Animals , Bile Acids and Salts/metabolism , Blotting, Northern , Blotting, Western , Chromatography, High Pressure Liquid , Cytosol/metabolism , DNA-Binding Proteins/metabolism , Enoyl-CoA Hydratase/chemistry , Hepatocyte Nuclear Factor 4 , Humans , Isomerases/chemistry , Liver/metabolism , Mice , Mice, Knockout , Models, Biological , Multienzyme Complexes/chemistry , Oxidoreductases/chemistry , Oxygen/chemistry , PPAR alpha/metabolism , Peroxisomal Bifunctional Enzyme , Peroxisomal Multifunctional Protein-2 , Peroxisomes/metabolism , Phosphoproteins/metabolism , Receptors, Cytoplasmic and Nuclear , Subcellular Fractions , Time Factors , Transcription Factors/metabolism , Up-Regulation
20.
J Clin Endocrinol Metab ; 89(11): 5661-8, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15531526

ABSTRACT

Expression of 17beta-hydroxysteroid dehydrogenases (17beta-HSDs) was compared between leiomyoma and myometrium. Cytosolic fractions from leiomyoma homogenate displayed 5-fold higher activity (estrone to estradiol), compared with surrounding myometrium (n = 6, P < 0.05), whereas microsomal fractions showed no difference. Oxidative activity (estradiol to estrone) did not differ between leiomyoma and myometrium. Levels of mRNA for 17beta-HSDs were then measured using real-time PCR techniques. Among the eight different types of 17beta-HSDs (types 1-5, 7, 8, and 10), type 1 was the only enzyme displaying differential expression between leiomyoma and myometrium. Mean concentration of type 1 17beta-HSD mRNA was 4-fold higher in leiomyoma than in surrounding myometrium (n = 20, P < 0.05). Type 1 transcript levels correlated significantly with reductive activity in individual samples (n = 6, P < 0.05). Northern blot analysis of leiomyoma and myometrium tissues detected 2.3- and 1.0-kb transcripts of type 1 enzyme, whereas the major 1.3-kb transcript for 17beta-HSD in placenta-derived JEG-3 cells was not detected. None of the factors increasing mRNA levels for type 1 enzyme in placenta increased mRNA levels in leiomyoma. These results indicate that leiomyoma tissues overexpress type 1 17beta-HSD, resulting in high conversion of estrone to estradiol. In situ expression of type 1 17beta-HSD may play a role in self-supported growth of leiomyoma cells.


Subject(s)
17-Hydroxysteroid Dehydrogenases/physiology , Estradiol/biosynthesis , Leiomyoma/metabolism , Uterine Neoplasms/metabolism , 17-Hydroxysteroid Dehydrogenases/genetics , Adult , Estrone/metabolism , Female , Gene Expression Regulation, Enzymologic , Humans , Middle Aged , RNA, Messenger/analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...